Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Iran J Pharm Res ; 19(1): 321-330, 2020.
Article in English | MEDLINE | ID: mdl-32922490

ABSTRACT

Marine macroalgae have attracted much attention in recent years as a valuable source of bioactive metabolites. The cytotoxic potential of the Laurencia papillosa red alga collected from the Lebanese coast has been investigated on human breast cancer cells MCF-7. The crude extract of Laurencia papillosa (L. papillosa) was fractionated by column chromatography using a series of increasingly polar solvents (methylene chloride, acetone and methanol). Cytotoxicity of the crude extract and fractions was determined by MTT assay in MCF-7 cells. Apoptosis was detected by annexin V/propidium iodide assay and by measurement of Bcl-2 expression. Flotillin-2 expression was examined using RT-qPCR and Western blot. The crude extract, and the fractions of CH2Cl2 and acetone exhibited a dose-dependent cytotoxic effect on MCF-7 cells. Apoptosis was specifically induced by one of the acetone fractions having the highest cytotoxicity. It has been demonstrated by an increase in late phase apoptotic cell populations, and a decrease in Bcl-2 anti-apoptotic marker expression on mRNA and protein levels in a dose- and time- dependent manner. Furthermore, this active fraction decreased Flotillin-2 expression associated with cancer progression. Our data suggest that L. papillosa is an important source of cytotoxic metabolites. Further studies are needed for the chemical characterization of the metabolite associated with observed biological activities.

2.
Lipids Health Dis ; 18(1): 168, 2019 Sep 02.
Article in English | MEDLINE | ID: mdl-31477154

ABSTRACT

BACKGROUND: The alteration of lipid metabolism in cancer cells is recognized as one of the most important metabolic hallmarks of cancer. Membrane rafts defined as plasma membrane microdomains enriched in cholesterol and sphingolipids serve as platforms for signaling regulation in cancer. The main purpose of this study was to evaluate the effect of the cholesterol metabolite, 4-cholesten-3-one, on lipid metabolism and membrane raft integrity in two breast cancer cell lines, MCF-7 and MDA-MB-231. Its ability to reduce cell viability and migration has also been investigated. METHODS: RT-qPCR was performed to evaluate the expression of enzymes involved in lipogenesis and cholesterol synthesis, and ABCG1 and ABCA1 transporters involved in cholesterol efflux. Its effect on cell viability and migration was studied using the MTT assay, the wound healing assay and the Transwell migration assay, respectively. The effect of 4-cholesten-3-one on membrane rafts integrity was investigated by studying the protein expression of flotillin-2, a membrane raft marker, and raft-enriched EGFR by western blot. RESULTS: Interestingly, we found that 4-cholesten-3-one treatment decreased mRNA expression of different enzymes including ACC1, FASN, SCD1 and HMGCR. We further demonstrated that 4-cholesten-3-one increased the expression of ABCG1 and ABCA1. We also found that 4-cholesten-3-one decreased the viability of MCF-7 and MDA-MB-231 cells. This effect was neutralized after treatment with LXR inverse agonist or after LXRß knockdown by siRNA. As a result, we also demonstrated that 4-cholesten-3-one disrupts membrane rafts and cell migration capacity. CONCLUSION: Our results show that 4-cholesten-3-one exerts promising antitumor activity by altering LXR-dependent lipid metabolism in breast cancer cells without increasing lipogenesis.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cholestenones/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Lipogenesis/drug effects , Liver X Receptors/genetics , Membrane Microdomains/drug effects , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism , Acetyl-CoA Carboxylase/genetics , Acetyl-CoA Carboxylase/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , ErbB Receptors/genetics , ErbB Receptors/metabolism , Fatty Acid Synthase, Type I/genetics , Fatty Acid Synthase, Type I/metabolism , Humans , Hydroxymethylglutaryl CoA Reductases/genetics , Hydroxymethylglutaryl CoA Reductases/metabolism , Lipogenesis/genetics , Liver X Receptors/metabolism , MCF-7 Cells , Membrane Microdomains/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Signal Transduction , Stearoyl-CoA Desaturase/genetics , Stearoyl-CoA Desaturase/metabolism , THP-1 Cells
3.
Cell Oncol (Dordr) ; 41(1): 13-24, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28993998

ABSTRACT

BACKGROUND: It has amply been documented that mammary tumor cells may exhibit an increased lipogenesis. Biliary acids are currently recognized as signaling molecules in the intestine, in addition to their classical roles in the digestion and absorption of lipids. The aim of our study was to evaluate the impact of lithocholic acid (LCA) on the lipogenesis of breast cancer cells. The putative cytotoxic effects of LCA on these cells were also examined. METHODS: The effects of LCA on breast cancer-derived MCF-7 and MDA-MB-231 cells were studied using MTT viability assays, Annexin-FITC and Akt phosphorylation assays to evaluate anti-proliferative and pro-apoptotic properties, qRT-PCR and Western blotting assays to assess the expression of the bile acid receptor TGR5 and the estrogen receptor ERα, and genes and proteins involved in apoptosis (Bax, Bcl-2, p53) and lipogenesis (SREBP-1c, FASN, ACACA). Intracellular lipid droplets were visualized using Oil Red O staining. RESULTS: We found that LCA induces TGR5 expression and exhibits anti-proliferative and pro-apoptotic effects in MCF-7 and MDA-MB-231 cells. Also, an increase in pro-apoptotic p53 protein expression and a decrease in anti-apoptotic Bcl-2 protein expression were observed after LCA treatment of MCF-7 cells. In addition, we found that LCA reduced Akt phosphorylation in MCF-7 cells, but not in MDA-MB-231 cells. We also noted that LCA reduced the expression of SREBP-1c, FASN and ACACA in both breast cancer-derived cell lines and that cells treated with LCA contained low numbers of lipid droplets compared to untreated control cells. Finally, a decrease in ERα expression was observed in MCF-7 cells treated with LCA. CONCLUSIONS: Our data suggest a potential therapeutic role of lithocholic acid in breast cancer cells through a reversion of lipid metabolism deregulation.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Breast Neoplasms/metabolism , Lipogenesis/drug effects , Lithocholic Acid/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Gene Expression/drug effects , Humans , Lipid Droplets/drug effects , MCF-7 Cells , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
4.
Anticancer Res ; 37(8): 4067-4073, 2017 08.
Article in English | MEDLINE | ID: mdl-28739689

ABSTRACT

BACKGROUND/AIM: Lipid rafts are cholesterol-enriched microdomains of the plasma membrane. Recent studies have underlined that their integrity is critical for cancer cell survival. Liver X receptor (LXR) has a central role in cellular cholesterol homeostasis and its stimulation inhibits proliferation of several cancer cell lines. This study investigated whether LXR could modulate lipid rafts integrity and consequently alter proliferation of the MCF-7 breast cancer cell line. MATERIALS AND METHODS: Effect of LXR agonist T0901317 on integrity of MCF-7 lipid rafts was examined by studying the expression of rafts marker flotillin-2 (FLOT2) and DHHC5, which palmitoylates FLOT2, and by studying the expression of phospho-Akt. RESULTS: We demonstrated that LXR stimulation decreases mRNA and protein expression of FLOT2 and DHHC5 in MCF-7 cells. LXR stimulation also reduces Akt phosphorylation and its localization at the plasma membrane. CONCLUSION: We showed, for the first time, that LXR regulates transcription of specific proteins of lipid rafts in a breast cancer model.


Subject(s)
Acyltransferases/genetics , Breast Neoplasms/drug therapy , Liver X Receptors/biosynthesis , Membrane Proteins/genetics , Apoptosis/drug effects , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cell Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Hydrocarbons, Fluorinated/administration & dosage , Liver X Receptors/agonists , Liver X Receptors/genetics , MCF-7 Cells , Membrane Microdomains/genetics , Membrane Microdomains/pathology , Phosphorylation , Sulfonamides/administration & dosage
5.
Anticancer Res ; 33(9): 3783-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24023310

ABSTRACT

BACKGROUND: Apolipoprotein E (APOE), a lipid transport protein that has a key role in the lipoprotein metabolism, is expressed by macrophages under the control of the transcription factor Liver X Receptor (LXR), an oxysterol-activated transcriptional factor involved in cholesterol metabolism. Recent work has shown that LXR agonists may inhibit breast cancer cell proliferation in vitro. We hypothesized that LXR-activated macrophages, and in particular secreted macrophagic APOE, may potentiate the effect of LXR agonists. Our goal was to evaluate the effect of APOE, secreted by THP-1 macrophages under the control of LXR, on MCF-7 cell proliferation, a model of breast cancer. MATERIALS AND METHODS: MCF-7 cells were incubated with supernatants from THP-1 cells previously treated with LXR agonists [T0901317 or 22(R)-hydroxycholestrol], or supernatants from THP-1 cells transfected with siRNA against APOE mRNA. RESULTS: Viability assays and cell death quantification showed that media from LXR-activated macrophages reduced cell proliferation and increased apoptosis of MCF-7 cells. Interestingly, the opposite effects were observed when MCF-7 cells wre treated with media from the siRNA APOE-mediated knock-down model. CONCLUSION: This study highlights the protective role of LXR-activated macrophages against breast cancer growth, and the implication of APOE protein in the anti-proliferative and pro-apoptotic effects observed.


Subject(s)
Apolipoproteins E/metabolism , Breast Neoplasms/metabolism , Cell Proliferation , Macrophages/metabolism , Orphan Nuclear Receptors/physiology , Apoptosis , Base Sequence , Breast Neoplasms/pathology , Culture Media, Conditioned , DNA Primers , Enzyme-Linked Immunosorbent Assay , Female , History, Ancient , Humans , Liver X Receptors , MCF-7 Cells , Orphan Nuclear Receptors/agonists , Orphan Nuclear Receptors/genetics , RNA Interference , RNA, Small Interfering , Real-Time Polymerase Chain Reaction
6.
Anticancer Res ; 32(7): 3007-13, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22753765

ABSTRACT

BACKGROUND: Liver X receptor (LXR) plays a key role in reverse cholesterol transport by inducing the expression of the ATP-binding cassette (ABC) transporters, implicated in cholesterol efflux. Recent data showed that LXR agonists inhibit the proliferation of multiple types of human cancer cells. However, whether these effects are related to cholesterol efflux has not yet been elucidated. MATERIALS AND METHODS: Effects of two LXR agonists (TO901317 and 22(R)-hydroxycholesterol [22(R)-HC]) on proliferation, apoptosis and cholesterol efflux were examined in MCF-7 breast cancer cells. RESULTS: Treatment with LXR agonists (TO901317 at 20 µM and 22(R)-HC at 2 µg/ml) inhibited proliferation and induced apoptosis of MCF-7 cells. Furthermore, LXR activation resulted in an increase in gene and protein levels of ABCG1 transporters and in cholesterol efflux to isolated high-density lipoprotein (HDL), without affecting the ABCA1/APOA-I mediated efflux. Under these conditions, a remarkable reduction of intracellular and membrane-associated cholesterol levels was observed. CONCLUSION: LXR activation in MCF-7 cells could deprive cells of cholesterol, required for their growth, by stimulating its efflux, resulting in the inhibition of cell proliferation and in stimulation of apoptosis.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cholesterol/metabolism , Hydroxycholesterols/pharmacology , Orphan Nuclear Receptors/agonists , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP-Binding Cassette Transporters/biosynthesis , ATP-Binding Cassette Transporters/genetics , Apoptosis/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Growth Processes/drug effects , Cell Line, Tumor , Female , Humans , Hydrocarbons, Fluorinated/pharmacology , Lipoproteins, HDL/metabolism , Liver X Receptors , Orphan Nuclear Receptors/metabolism , Sulfonamides/pharmacology
7.
J Cell Biochem ; 106(4): 659-65, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19160412

ABSTRACT

An unusual sterolic mixture (82.3% of 24-isopropylated sterols) and its major component, 24-isopropylcholesterol, isolated from a marine sponge, Ciocalypta sp. (Halichondriidae), reduce cholesterol uptake, basolateral secretion and ACAT-2 mRNA expression and increase the expression of ABCA1 mRNA in Caco-2 cells. The decreases of cholesterol uptake and secretion induced by 24-isopropylcholesterol alone were more than that of both the sterolic mixture and beta-sitosterol. These data add a new sterol, 24-isopropylcholesterol, to sterols that may reduce intestinal cholesterol absorption.


Subject(s)
Cholesterol/metabolism , Sterols/pharmacology , Animals , Caco-2 Cells , Dehydrocholesterols/pharmacology , Humans , Intestinal Absorption/drug effects , Porifera , RNA, Messenger/analysis , Sitosterols/pharmacology , Sterol O-Acyltransferase/genetics , Sterol O-Acyltransferase 2
8.
Metabolism ; 55(7): 980-8, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16784973

ABSTRACT

Epidemiological data suggest a link between chronic inflammation condition and atherosclerosis. Infection and inflammation can also impair lipoprotein metabolism and produce a wide variety of changes in plasma concentrations of lipids and lipoproteins. Twenty-one patients with inflammatory bowel diseases (IBDs) and 28 healthy subjects were recruited. Serum concentrations of lipids, lipoproteins, apolipoproteins, leptin, ghrelin, and inflammation markers (C-reactive protein and serum amyloid A) were measured, and subjects' lipoproteins were characterized. The ability of patients with serum IBD to efflux free cell cholesterol was measured. Serum cholesterol, high-density lipoprotein cholesterol, apolipoprotein (apo) A-I, apoC-II, apoC-III bound to apoB, phospholipid, and phospholipids not bound to apoB levels were significantly lower, whereas serum triglyceride, serum amyloid A, and C-reactive protein levels were significantly higher in patients with active IBD. Apolipoprotein A-I immunoreactivity (pre-beta small particles and small alpha-high-density lipoprotein particles) is decreased in patients with IBD. In contrast, apoE immunoreactivity (slow/small apoE containing lipoprotein particles [LpE particle]) increased in these patients. The efflux capacity of serum from patients with IBD using [(3)H]-cholesterol-labeled Fu5AH cells was reduced (P < .005). Our results demonstrate that, in subjects with active IBD, inflammation leads to alterations in lipid, apolipoprotein, and lipoprotein profiles and reduced cholesterol efflux. These changes are similar to those proposed to promote atherogenesis and may contribute to the development of cardiovascular events.


Subject(s)
Apolipoproteins/blood , Cholesterol/metabolism , Inflammatory Bowel Diseases/blood , Lipids/blood , Lipoproteins/blood , Adult , Cells, Cultured , Electrophoresis, Gel, Two-Dimensional , Female , Humans , Male , Middle Aged , Regression Analysis , Serum Amyloid A Protein/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...